Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Chem Biol Interact ; 350: 109707, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34656558

ABSTRACT

The organochlorine pesticide dicofol (DCF), a persistent organic pollutant, is used as acaricide worldwide. Considering its large consumption in the agriculture sector and potential toxic effects such as endocrine disruption, carcinogenicity, and environmental persistence are detrimental to human health. To take an extensive evaluation of its potential toxicity, the current study was aimed to explore the binding mechanism and adverse effect of DCF on human serum albumin (HSA) by using an array of biophysical techniques (UV-visible, fluorescence, 3D fluorescence, and circular dichroism spectroscopy), isothermal titration calorimetric (ITC), computational methods and biochemical approaches. Fluorescence quenching and UV-Visible spectra of the HSA-DCF system confirmed static quenching mechanism and complex formation between HSA and DCF. The thermodynamics results from ITC revealed DCF-HSA interaction was exothermic and spontaneous and involved hydrophobic interactions and hydrogen bonding. The esterase activity of HSA displayed constant Vmax and elevated Km values confirming DCF-HSA competitive interaction. Circular dichroism spectra results revealed structural changes in HSA protein on interaction with DCF. Furthermore, molecular-specific site marker and molecular modelling results affirmed that the binding Site of DCF is Site I of HSA. A significant carbonyl content level in DCF-HSA system suggested protein structure damage. This work is likely to add a better understanding of DCF toxicity in human health and helpful in fortifying the check on food safety.


Subject(s)
Dicofol/pharmacokinetics , Dicofol/toxicity , Persistent Organic Pollutants/pharmacokinetics , Persistent Organic Pollutants/toxicity , Pesticides/pharmacokinetics , Pesticides/toxicity , Serum Albumin, Human/drug effects , Serum Albumin, Human/metabolism , Binding Sites , Circular Dichroism , Dicofol/chemistry , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , In Vitro Techniques , Kinetics , Molecular Docking Simulation , Persistent Organic Pollutants/chemistry , Pesticides/chemistry , Protein Binding , Serum Albumin, Human/chemistry , Spectrometry, Fluorescence , Spectrum Analysis , Thermodynamics
2.
Food Funct ; 12(8): 3692-3704, 2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33900309

ABSTRACT

Monascin (MS) and ankaflavin (AK), as typical yellow lipid-soluble pigments identified from Monascus-fermented products, have been confirmed to possess diverse biological activities such as anti-oxidation, reversing diabetes, and anti-atherosclerosis, and have received increasing attention in recent years. Certainly Monascus-fermented product with a high content of MS/AK is also a concern. The current work explored interactions between MS/AK and human serum albumin (HSA) as well as their influence on the anti-oxidant properties of MS/AK. Moreover, the anti-glycation potential of Monascus-fermented products rich in MS and AK (denoted as Mps) was assessed. The results showed that the fluorescence emission of HSA was quenched by MS/AK through a static quenching mechanism, and MS-HSA and AK-HSA complexes were mainly formed by van der Waals forces and hydrophobic interactions, but AK showed a higher binding affinity than MS. Although the DPPH radical-scavenging abilities of MS-HSA and AK-HSA complexes declined, Mps significantly reduced the formation of fructosamine, α-dicarbonyl compounds and advanced glycation end products (AGEs) in the in vitro glycation model (HSA-glucose). Notably, approximately 80% of fluorescent-AGEs were suppressed by Mps at a concentration of 0.95 mg mL-1, while aminoguanidine (AG, a reference standard) caused only 65% decrease at the same concentration. Although radical scavenging and metal chelating activities could justify the observed anti-glycation activity of Mps, in-depth research on the structures of other functional compounds present in Mps except MS/AK and reaction mechanisms should be performed. Overall, the present study proved that Mps would be promising sources of food-based anti-glycation agents because of their superior inhibitory effect on AGEs.


Subject(s)
Antioxidants/pharmacology , Flavins/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , Monascus , Antioxidants/chemistry , Flavins/chemistry , Glycosylation/drug effects , Heterocyclic Compounds, 3-Ring/chemistry , Humans , Serum Albumin, Human/drug effects
3.
An Acad Bras Cienc ; 92(1): e20190819, 2020.
Article in English | MEDLINE | ID: mdl-32491127

ABSTRACT

Herbal-flavonoids (HF) as polyphenolic secondary metabolites are taken in the daily diet to join in many metabolic processes in the human organism. Anti-proliferative activities and human serum albumin (HSA) binding capacities of herbal-flavonoids namely 7,5'-dimethoxyisoetin (HF1), homoorientin-6''-4-O-methyl-myo-inositol (HF2), (2R, 3R)-(+)-dihydrokaempferol-7,4'-dimethylether (HF3), eriodictyol-7,4'-dimethylether (HF4) and flavonoids isoorientin (HF5) and genkwanin (HF6) were investigated. Anti-proliferative activities were determined by the xCELLigence system by treatment with human prostate (PC3) and cervical cancer (HeLa) cells. The binding capacities were studied by two-dimensional (2D-FL) and three-dimensional (3D-FL) fluorescence spectroscopy. HeLa and PC3 cell lines were treated with flavonoids at 10, 50 and 100 µg/mL concentrations over a 48 hour period. Stable anti-proliferative efficacy plots were obtained for tested flavonoids. From the flavonoids, HF3 and HF4 showed the strongest anti-proliferative effect against PC3 and HeLa cell line. HF1 and HF2 exhibited the strongest binding capacity to the HSA corresponding to Kb values of 3.81 x 104 M-1 and 6.00 x 104 M-1, respectively. The studies revealed that the flavonoids form the basis of in vivo preclinical studies as important nutraceuticals of the daily diet, as well as modelled in medical and pharmacological applications.


Subject(s)
Cell Proliferation/drug effects , Flavonoids/pharmacology , Serum Albumin, Human/drug effects , Flavonoids/chemistry , HeLa Cells , Humans , Spectrometry, Fluorescence
4.
Spectrochim Acta A Mol Biomol Spectrosc ; 226: 117652, 2020 Feb 05.
Article in English | MEDLINE | ID: mdl-31654902

ABSTRACT

Transient absorption spectroscopy in combination with in silico methods has been employed to study the interactions between human serum albumin (HSA) and the anti-psychotic agent chlorpromazine (CPZ) as well as its two demethylated metabolites (MCPZ and DCPZ). Thus, solutions containing CPZ, MCPZ or DCPZ and HSA (molar ligand:protein ratios between 1:0 and 1:3) were submitted to laser flash photolysis and the ΔAmax value at λ = 470 nm, corresponding to the triplet excited state, was monitored. In all cases, the protein-bound ligand exhibited higher ΔAmax values measured after the laser pulse and were also considerably longer-lived than the non-complexed forms. This is in agreement with an enhanced hydrophilicity of the metabolites, due to the replacement of methyl groups with H that led to a lower extent of protein binding. For the three compounds, laser flash photolysis displacement experiments using warfarin or ibuprofen indicated Sudlow site I as the main binding site. Docking and molecular dynamics simulation studies revealed that the binding mode of the two demethylated ligands with HSA would be remarkable different from CPZ, specially for DCPZ, which appears to come from the different ability of their terminal ammonium groups to stablish hydrogen bonding interactions with the negatively charged residues within the protein pocket (Glu153, Glu292) as well as to allocate the methyl groups in an apolar environment. DCPZ would be rotated 180° in relation to CPZ locating the aromatic ring away from the Sudlow site I of HSA.


Subject(s)
Chlorpromazine/chemistry , Chlorpromazine/pharmacokinetics , Serum Albumin, Human/chemistry , Serum Albumin, Human/metabolism , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antipsychotic Agents/chemistry , Antipsychotic Agents/pharmacokinetics , Binding Sites , Carbazoles/chemistry , Carbazoles/pharmacokinetics , Chlorpromazine/analogs & derivatives , Chlorpromazine/pharmacology , Drug Interactions , Humans , Hydrogen Bonding , Inactivation, Metabolic , Methylation , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding/drug effects , Serum Albumin, Human/drug effects , Spectrophotometry, Ultraviolet , Stereoisomerism
5.
Inorg Chem ; 58(23): 16154-16170, 2019 Dec 02.
Article in English | MEDLINE | ID: mdl-31721562

ABSTRACT

In this study, two new bis-cyclometalated Pt(II) complexes, [Pt(C^N)(S^N)] [S^N = deprotonated 6-mercaptopurine (6-MP) and C^N = deprotonated 2-phenylpyridine (ppy), 2a; C^N = deprotonated benzo[h]quinoline (bhq), 2b], are synthesized by the reaction of [PtR(SMe2)(C^N)] (R = Me or p-MeC6H4) with 1 equiv of 6-mercaptopurine (6-HMP) at room temperature. The complexes are fully characterized using 1H and 13C NMR spectroscopies, electrospray ionization mass spectrometry, and elemental analysis. Biomolecular interaction of complex 2a with human serum albumin (HSA) is studied by fluorescence, UV-vis, and circular dichroism (CD) spectroscopies. The binding constants (Kb) and number of binding sites (n) are evaluated using the Stern-Volmer equation. The intrinsic fluorescence of protein is quenched by a static quenching mechanism, with a binding constant of Kb ∼ 105 reflecting a high affinity of complex 2a for HSA. The thermodynamic parameters (ΔH°, ΔG°, and ΔS°) indicate that the interaction is a spontaneous process and hydrophobic forces play a main role in the reaction. The displacement experiments demonstrate that the reactive binding sites of HSA to complex 2a are mainly located within its hydrophobic cavity in subdomain IIA (site I). Synchronous fluorescence spectra reveal that complex 2a affected the microenvironment of tryptophan-214 residues in subdomain IIA of HSA. In the case of interaction of complex 2b and HSA, because of overlapping of the emission spectra of complex 2b with HSA, chemometric approaches are applied. The results indicate significant interaction between the tryptophan residue of HSA and complex 2b. Moreover, the binding of Pt(II) complexes 2a and 2b causes a reduction of the α-helix content of HSA, as obtained by far-UV CD spectroscopy. The average binding distance (r) between Pt(II) complexes and HSA is obtained by Förster's resonance energy-transfer theory. Also, a molecular docking simulation reveals that π-π-stacking and hydrophobic interactions between these complexes and HSA are significant. Furthermore, the interactions of platinum complexes, 2, with calf-thymus DNA (CT-DNA) are investigated. The UV-vis results and ethidium bromide competitive studies support an intercalative interaction of both Pt(II) complexes with DNA. The new complexes 2 are also screened for anticancer activities. The results show that complexes 2 exhibit significant anticancer activity against the K562 (chronic myelogenous leukemia) cell line.


Subject(s)
Antineoplastic Agents/pharmacology , DNA/drug effects , Mercaptopurine/pharmacology , Organoplatinum Compounds/pharmacology , Serum Albumin, Human/drug effects , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Binding Sites/drug effects , Cell Proliferation/drug effects , DNA/chemistry , Density Functional Theory , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , K562 Cells , Mercaptopurine/chemistry , Molecular Docking Simulation , Molecular Structure , Organoplatinum Compounds/chemical synthesis , Organoplatinum Compounds/chemistry , Serum Albumin, Human/chemistry , Structure-Activity Relationship , Thermodynamics
6.
Molecules ; 24(9)2019 May 05.
Article in English | MEDLINE | ID: mdl-31060332

ABSTRACT

Hair-coloring products include permanent, semi-permanent and temporary dyes that vary by chemical formulation and are distinguished mainly by how long they last. Domestic temporary hair dyes, such as fuchsin basic, basic red 2 and Victoria blue B, are especially popular because of their cheapness and facile applications. Despite numerous studies on the relationship between permanent hair dyes and disease, there are few studies addressing whether these domestic temporary hair dyes are associated with an increased cancer risk. Herein, to ascertain the bio-safety of these temporary hair dyes, we comparatively studied their percutaneous absorption, hemolytic effect and cytotoxic effects in this paper. Furthermore, to better understand the risk of these dyes after penetrating the skin, experimental and theoretical studies were carried out examining the interactions between the dyes and serum albumins as well as calf thymus (CT)-DNA. The results showed that these domestic temporary hair dyes are cytotoxic with regard to human red blood cells and NIH/3T3 cell lines, due to intense interactions with bovine serum albumin (BSA)/DNA. We conclude that the temporary hair dyes may have risk to human health, and those who use them should be aware of their potential toxic effects.


Subject(s)
Erythrocytes/cytology , Hair Dyes/adverse effects , NIH 3T3 Cells/cytology , Rosaniline Dyes/adverse effects , Animals , Cattle , Cell Survival/drug effects , DNA/drug effects , Erythrocytes/drug effects , Hair Dyes/chemistry , Hair Dyes/pharmacokinetics , Hemolysis , Humans , Mice , Molecular Docking Simulation , NIH 3T3 Cells/drug effects , Phenazines/adverse effects , Phenazines/chemistry , Phenazines/pharmacokinetics , Rosaniline Dyes/chemistry , Rosaniline Dyes/pharmacokinetics , Serum Albumin, Human/drug effects , Swine
7.
J Med Chem ; 62(10): 5063-5079, 2019 05 23.
Article in English | MEDLINE | ID: mdl-31074269

ABSTRACT

A common source of false positives in drug discovery is ligand self-association into large colloidal assemblies that nonspecifically inhibit target proteins. However, the mechanisms of aggregation-based inhibition (ABI) and ABI-attenuation by additives, such as Triton X-100 (TX) and human serum albumin (HSA), are not fully understood. Here, we investigate the molecular basis of ABI and ABI-attenuation through the lens of NMR and coupled thermodynamic cycles. We unexpectedly discover a new class of aggregating ligands that exhibit negligible interactions with proteins but act as competitive sinks for the free inhibitor, resulting in bell-shaped dose-response curves. TX attenuates ABI by converting inhibitory, protein-binding aggregates into nonbinding coaggregates, whereas HSA minimizes nonspecific ligand interactions by functioning as a reservoir for free inhibitor and preventing self-association. Hence, both TX and HSA are useful tools to minimize false positives arising from nonspecific binding but at the cost of potentially introducing false negatives due to suppression of specific interactions.


Subject(s)
Drug Discovery/methods , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Binding Sites , Buffers , Dose-Response Relationship, Drug , False Positive Reactions , Humans , Light , Magnetic Resonance Spectroscopy , Octoxynol/pharmacology , Scattering, Radiation , Serum Albumin, Human/chemistry , Serum Albumin, Human/drug effects , Spectrometry, Mass, Electrospray Ionization , Surface Plasmon Resonance , Thermodynamics
8.
Article in English | MEDLINE | ID: mdl-30496974

ABSTRACT

Albumin is a new biomarker of organophosphorus compounds (OPs) and nerve agents (OPNAs) for retrospective verification. Recent studies on OPs adducts show that amino acid residues can covalently bind to OPs and OPNAs. In this article, after being incubated with soman, sarin, cyclosarin, VX, ethyl tabun, and propyl tabun, human serum albumin (HSA) is analyzed by quadrupole-Orbitrap mass spectrometer (Q Exactive LC-MS/MS). In addition to the three known phosphonylated sites, six new sites modified by OPNAs are detected. To identify the most reactive residue, we calculate the area ratio of the modified peptides to the whole peptides. The result demonstrates that tyrosine 263 (Y263) in peptide Y263ICENQDSISSK, which has been poisoned with six kinds of nerve agents, possesses the highest reactivity. The structure characteristics based on molecular simulation provide a theoretical evidence for the reactivity of the nine binding sites. It suggests that Y263 also has the potential to be used as a biomarker to detect OPNAs exposure, and the presented Q Exactive LC-MS/MS method might be of relevance for the verification of new phosphonylated sites.


Subject(s)
Nerve Agents/poisoning , Serum Albumin, Human/chemistry , Serum Albumin, Human/drug effects , Biomarkers/analysis , Biomarkers/chemistry , Humans , Models, Chemical , Serum Albumin, Human/analysis , Tandem Mass Spectrometry , Tyrosine/chemistry
9.
J Biomol Struct Dyn ; 37(11): 2776-2788, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30101645

ABSTRACT

Green tea has attracted great interest as a cancer prevention agent. Interactions of tea polyphenols with serum albumin may influence the efficacy of drugs. The interactions of (-)-epigallocatechin-3-gallate (EGCG), (-)-epicatechin-3-gallate (ECG), and tegafur (TF) alone or in combination with human serum albumin (HSA) at pH 7.4 and different temperatures were investigated by spectroscopic methods, isothermal titration calorimetry (ITC), and molecular docking. The binding affinities to HSA were ranked in the order of EGCG > ECG > TF, and the interactions were spontaneous and exothermic. Ternary system studies showed that the presence of one component hindered the binding of another component to HSA. The secondary structures of HSA were slightly altered in the presence of the ligands. Site marking experiments and molecular docking showed that EGCG and ECG mainly bound to subdomain IIA and ΙΙΙA while TF bound to subdomain ΙΙA and ΙB. Results indicated that the existence of ECG and EGCG would influence the binding of TF to HSA and can increase the free concentration of TF. Obtained results would provide beneficial information about possible interference upon simultaneous co-administration of the tea components and drugs. Communicated by Ramaswamy H. Sarma.


Subject(s)
Calorimetry/methods , Catechin/analogs & derivatives , Molecular Docking Simulation , Serum Albumin, Human/metabolism , Spectrometry, Fluorescence/methods , Tegafur/metabolism , Antioxidants/pharmacology , Binding Sites , Biophysical Phenomena , Catechin/pharmacology , Humans , Protein Binding , Protein Conformation , Serum Albumin, Human/chemistry , Serum Albumin, Human/drug effects , Tegafur/chemistry , Thermodynamics
10.
J Biomol Struct Dyn ; 37(11): 2789-2800, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30052136

ABSTRACT

In this study, Farnesiferol C was introduced as an anti-colon cancer agent. Its cytotoxicity was investigated on two cancer cell lines, HCT116 and CT26, and mesenchymal stem cells (MSCs) as normal cells employing MTT assay. Moreover, Farnesiferol C interactions with ct-DNA and HSA were investigated by various techniques. The IC50 values of Farnesiferol C on HCT116 and CT26 cells were 42 and 46 µM, respectively, while its IC50 value on MSCs cells was 92 µM, indicating that Farnesiferol C was more efficacious against cancer cell lines than normal cells. DNA competitive binding studies, viscosity and zeta potential measurements confirmed that Farnesiferol C bound to DNA through intercalation binding. HSA binding investigations revealed that there were two different binding sites for Far C on HSA with higher binding affinity in site 2 compared to site 1. Furthermore, Farnesiferol C could bind to HSA and quench its intrinsic fluorescence in a static quenching mechanism, with a distance of 2.54 nm. Competitive binding in the presence of warfarin and ibuprofen was carried out and the resulting quenching constant was strongly changed in the presence of warfarin. Consequently, Farnesiferol C most probably will be located within sub-domain IIA. In this study, molecular modeling buttressed and confirmed our laboratory results. Conclusively, we proposed that DNA is an appropriate target for Farnesiferol C. Therefore, Farnesiferol C and its semisynthetic analogues can be one of the priority innovations in research on anticancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Coumarins/pharmacology , DNA/metabolism , Ferula/chemistry , Mesenchymal Stem Cells/drug effects , Serum Albumin, Human/metabolism , Animals , Antineoplastic Agents/chemistry , Binding Sites , Binding, Competitive , Cell Proliferation , Cells, Cultured , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Coumarins/chemistry , DNA/chemistry , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Rats , Serum Albumin, Human/chemistry , Serum Albumin, Human/drug effects
11.
Curr Top Med Chem ; 18(26): 2230-2238, 2018.
Article in English | MEDLINE | ID: mdl-30569859

ABSTRACT

Poor profiles of potential drug candidates, including pharmacokinetic properties, have been acknowledged as a significant hindrance to the development of modern therapeutics. Contemporary drug discovery and development would be incomplete without the aid of molecular modeling (in-silico) techniques, allowing the prediction of pharmacokinetic properties such as clearance, unbound fraction, volume of distribution and bioavailability. As with all models, in-silico approaches are subject to their interpretability, a trait that must be balanced with accuracy when considering the development of new methods. The best models will always require reliable data to inform them, presenting significant challenges, particularly when appropriate in-vitro or in-vivo data may be difficult or time-consuming to obtain. This article seeks to review some of the key in-silico techniques used to predict key pharmacokinetic properties and give commentary on the current and future directions of the field.


Subject(s)
Ibuprofen/pharmacokinetics , Models, Molecular , Warfarin/pharmacokinetics , Administration, Oral , Humans , Ibuprofen/administration & dosage , Ibuprofen/chemistry , Quantitative Structure-Activity Relationship , Serum Albumin, Human/chemistry , Serum Albumin, Human/drug effects , Warfarin/administration & dosage , Warfarin/chemistry
12.
Pediatr Nephrol ; 33(12): 2299-2309, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30091061

ABSTRACT

BACKGROUND: Dyslipidemia is an important cardiovascular risk factor in steroid-resistant nephrotic syndrome (SRNS). Efficacy of statins for treatment of hyperlipidemia in children with SRNS is unclear. METHODS: This prospective, randomized, double-blind, placebo-controlled, parallel-group clinical trial enrolled 30 patients with SRNS, aged 5-18 years, with serum low-density lipoprotein cholesterol (LDL-C) levels between 130 and 300 mg/dl, to receive a fixed dose of atorvastatin (n = 15, 10 mg/d) or placebo (n = 15) by block randomization in a 1:1 ratio. Primary outcome was change in serum LDL-C at 12 months. Change in levels of other lipid fractions, carotid intima-media thickness (cIMT), flow-mediated dilation (FMD) of the brachial artery, and adverse events were also evaluated. RESULTS: At the end of 12 months, atorvastatin was not superior to placebo in reducing plasma LDL-C levels, median percentage reduction 15.8% and 9.5% respectively, in atorvastatin and placebo arms (n = 14 in each; P = 0.40). Apolipoprotein B levels significantly declined with atorvastatin in modified intention-to-treat analysis (P = 0.01) but not in the per-protocol analysis. There was no significant effect on other lipid fractions, cIMT and FMD. Adverse events were similar between groups. Change in serum albumin was negatively associated with change in serum LDL-C, very low-density lipoprotein cholesterol, total cholesterol, triglyceride, and apolipoprotein B (P < 0.001), irrespective of receiving atorvastatin, age, gender, body mass index, and serum creatinine. CONCLUSIONS: Atorvastatin, administered at a fixed daily dose of 10 mg, was not beneficial in lowering lipid levels in children with SRNS; rise in serum albumin was associated with improvement in dyslipidemia.


Subject(s)
Atherosclerosis/prevention & control , Atorvastatin/administration & dosage , Dyslipidemias/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Nephrotic Syndrome/complications , Adolescent , Atherosclerosis/blood , Atherosclerosis/etiology , Atorvastatin/adverse effects , Carotid Arteries/diagnostic imaging , Carotid Arteries/drug effects , Carotid Intima-Media Thickness , Child , Cholesterol, LDL/blood , Cholesterol, LDL/drug effects , Double-Blind Method , Dyslipidemias/blood , Dyslipidemias/etiology , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Male , Nephrotic Syndrome/blood , Prospective Studies , Serum Albumin, Human/analysis , Serum Albumin, Human/drug effects , Treatment Outcome
13.
Medicine (Baltimore) ; 97(27): e11282, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29979395

ABSTRACT

BACKGROUND: The present study aims to comprehensively determine the efficacy of different therapy regimens based on Tripterygium wilfordii Hook F (TwHF) for patients with primary nephrotic syndrome (PNS) using network meta-analysis method. METHODS: Seven electronic databases were searched to identify randomized controlled trials (RCTs) that compared the differences between different therapy regimens based on TwHF for patients with PNS. The risk of bias in included RCTs was evaluated according to the Cochrane Handbook version 5.2.0. Network meta-analysis was performed to compare different regimens. Primary outcomes were complete remission rate and total remission rate. The secondary outcomes were hr urinary protein excretion, serum albumin, serum creatinine, and urea nitrogen. Data analysis was performed using R software. RESULTS: A total of 40 studies involving 2846 patients with PNS were included. Compared with prednisone, the improvement in total remission rate and complete remission rate was associated with TwHF alone (odds ratio [OR] = 4.80, 95% credible intervals [CrI]: 2.20-10.00; OR = 6.30, 95% CrI: 2.90-13.00, respectively), TwHF+prednisone (OR = 2.10, 95% CrI: 1.30-3.50; OR = 2.40, 95% CrI: 1.50-3.80, respectively), TwHF+CPA (OR = 12.00, 95% CrI: 1.10-150.00; OR = 16.00, 95% CrI: 1.60-170.00, respectively), and TwHF+Cyclosporine A (OR = 28.00, 95% CrI: 3.20-250.00; OR = 35.00, 95% CrI: 4.50-270.00, respectively). Compared with TwHF alone, TwHF+prednisone showed less benefit in improving total remission rate and complete remission rate (OR = 0.44, 95% CrI: 0.21-0.91; OR = 0.38, 95% CrI: 0.19-0.77, respectively). TwHF alone, TwHF+prednisone could significantly reduce hr urinary protein excretion (MD = -0.69, 95% CrI: -1.30 to -0.14; MD = -1.00, 95% CrI: -1.90 to -0.14, respectively) and increase serum albumin (MD = 5.90, 95% CrI: 2.50-9.30; MD = 3.40, 95% CrI: 1.30-5.50, respectively) when compared to prednisone alone. TwHF alone showed significant reduction in serum creatinine when compared to CPA (MD = -19.00, 95% CrI: -37.00 to -0.56). CONCLUSIONS: TwHF alone, the addition TwHF to prednisone showed more benefit in improving total and complete remission rate, hr urinary protein excretion, serum albumin, and serum creatinine.


Subject(s)
Immunosuppressive Agents/therapeutic use , Medicine, Chinese Traditional/methods , Nephrotic Syndrome/drug therapy , Phytotherapy/methods , Tripterygium/drug effects , Glucocorticoids/therapeutic use , Humans , Kidney Function Tests , Network Meta-Analysis , Prednisone/therapeutic use , Serum Albumin, Human/drug effects , Treatment Outcome
14.
J Mol Recognit ; 31(8): e2713, 2018 08.
Article in English | MEDLINE | ID: mdl-29656610

ABSTRACT

Fipronil is a broad-spectrum pesticide widely used in agriculture, horticulture, and forestry. Because fipronil can cause a variety of toxic effects in animals and humans, its use is authorized as a pesticide in veterinary medicinal products for pets, but not for the treatment of livestock animals whose products are intended for consumption. Recently, however, the presence of fipronil residues has been detected in the eggs and meat of layer hens from farms located in different European countries. Given the relevance of fipronil toxicity for human health, it is important to gain information concerning its fate in the human body, including its binding mode to human serum albumin (HSA), the most abundant protein in plasma. Here, the inhibition of heme-Fe(III) binding to the fatty acid site 1 (FA1) of HSA by fipronil is reported. Docking simulations support functional data, indicating that the FA1 site is the preferential cleft for fipronil recognition by HSA. The affinity of fipronil for HSA (Kf  = 1.9 × 10-6  M, at pH 7.3, and 20.0°C) may be relevant in vivo. Indeed, HSA could play a pivotal role in fipronil transport and scavenging, thus reducing the pesticide-free plasmatic levels, with consequent reduced systemic toxicity. In turn, fipronil binding to the FA1 site of HSA could impair the recognition of endogenous and exogenous molecules.


Subject(s)
Pesticides/chemistry , Protein Conformation/drug effects , Pyrazoles/chemistry , Serum Albumin, Human/chemistry , Allosteric Regulation/drug effects , Animals , Binding Sites/drug effects , Chickens , Fatty Acids , Humans , Kinetics , Pesticides/adverse effects , Pesticides/pharmacology , Protein Binding/drug effects , Pyrazoles/adverse effects , Pyrazoles/pharmacology , Serum Albumin, Human/drug effects
15.
J Mol Recognit ; 31(8): e2715, 2018 08.
Article in English | MEDLINE | ID: mdl-29630759

ABSTRACT

In the present study, the interaction of human serum albumin (HSA) with some cardiovascular drugs (CARs) under physiological conditions was investigated via the fluorescence spectroscopic and Fourier transform infrared spectroscopy. The CAR included Captopril, Timolol, Propranolol, Atenolol, and Amiodarone. Cardiovascular drugs can effectively quench the endogenous fluorescence of HSA by static quenching mechanism. The fluorescence quenching of HSA is mainly caused by complex formation of HSA with CAR. The binding reaction of CAR with HSA can be concluded that hydrophobic and electrostatic interactions are the main binding forces in the CAR-HSA system. The results showed that CAR strongly quenched the intrinsic fluorescence of HSA through a static quenching procedure, and nonradiation energy transfer happened within molecules. Fourier transform infrared spectroscopy absorption studies showed that the secondary structure was changed according to the interaction of HSA and CAR. The binding reaction of CAR with HSA can be concluded that hydrophobic and electrostatic interactions are the main binding forces in the CAR-HSA system. The results obtained herein will be of biological significance in pharmacology and clinical medicines.


Subject(s)
Cardiovascular Agents/chemistry , Protein Binding/drug effects , Serum Albumin, Human/chemistry , Amiodarone/chemistry , Amiodarone/pharmacology , Atenolol/chemistry , Atenolol/pharmacology , Captopril/chemistry , Captopril/pharmacology , Cardiovascular Agents/therapeutic use , Humans , Hydrophobic and Hydrophilic Interactions/drug effects , Propranolol/chemistry , Propranolol/pharmacology , Serum Albumin, Human/drug effects , Spectrometry, Fluorescence , Spectroscopy, Fourier Transform Infrared , Static Electricity , Timolol/chemistry , Timolol/pharmacology
16.
Environ Sci Pollut Res Int ; 25(18): 17735-17748, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29671232

ABSTRACT

Some tetrahydroisoindoline-1,3-dione derivatives (TDDs) possess potent herbicidal activity. To assess possible impacts of TDDs on humans, the interactions between TDDs and human serum albumin (HSA) were evaluated with steady-state and time-resolved fluorescence spectroscopy, synchronous fluorescence spectroscopy, Fourier transform-infrared spectroscopy, and circular dichroism spectroscopy. The thermodynamic data obtained at temperatures of 298, 307, and 316 K indicate that TDDs spontaneously bind to HSA and thus form a TDD-HSA complex. The conformation and secondary structure of HSA are changed, and the intrinsic fluorescence of HSA is statically quenched by TDDs. Moreover, the TDD-HSA complex is formed primarily through electrostatic interactions and has only one binding site on HSA. A competitive ligand-binding assay revealed that site II (subdomain IIIA) displays the greatest affinity for TDDs. In addition, an acute toxicity bioassay showed no zebrafish mortality upon exposure to 4000 µg L-1 of TDDs. This work is helpful for understanding interactions between TDDs and HSA.


Subject(s)
Herbicides/chemistry , Herbicides/toxicity , Indoles/toxicity , Serum Albumin, Human/drug effects , Binding Sites , Fluorescence , Herbicides/metabolism , Humans , Indoles/chemistry , Indoles/metabolism , Ligands , Protein Structure, Secondary , Serum Albumin, Human/metabolism , Spectrometry, Fluorescence , Spectroscopy, Fourier Transform Infrared , Static Electricity , Thermodynamics
17.
Article in English | MEDLINE | ID: mdl-29324720

ABSTRACT

The herbicide atrazine is widely used across the globe, which is a great concern. To investigate its potential toxicity in the human body, human serum albumin (HSA) was selected as a model protein. The interaction between atrazine and HSA was investigated using steady-state fluorescence spectroscopy, synchronous fluorescence spectroscopy, UV-Vis spectroscopy, three-dimensional (3D) fluorescence spectroscopy and circular dichroism (CD) spectroscopy. The intrinsic fluorescence of HSA was quenched by the atrazine through a static quenching mechanism. Fluorescence spectra at two excitation wavelengths (280 and 295 nm) showed that the fluorescence quenched in HSA was mainly contributed to by tryptophan residues. In addition, the atrazine bound to HSA, which induced changes in the conformation and secondary structure of HSA and caused an energy transfer. Thermodynamic parameters revealed that this binding is spontaneous. Moreover, electrostatic interactions play a major role in the combination of atrazine and HSA. One atrazine molecule can only bind to one HSA molecule to form a complex, and the atrazine molecule is bound at site II (subdomain IIIA) of HSA. This study furthers the understanding of the potential effects posed by atrazine on humans at the molecular level.


Subject(s)
Atrazine/toxicity , Herbicides/toxicity , Serum Albumin, Human/drug effects , Atrazine/metabolism , Circular Dichroism , Fluorescence , Herbicides/metabolism , Humans , Protein Binding , Serum Albumin , Serum Albumin, Human/metabolism , Spectrometry, Fluorescence , Static Electricity , Thermodynamics
18.
Toxicol Lett ; 293: 31-37, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-29277572

ABSTRACT

The banned chemical warfare agent sulfur mustard (SM) still represents a serious threat to civilians and military personnel. Therefore, identification of antidotes and scavengers is of high concern. One promising substance is glutathione (GSH). GSH is known to mitigate symptoms of SM poisoning in vitro and in vivo. However, the mechanism of action remains unclear with respect to physiological impact as well as chemical scavenging by reaction between GSH and SM. Therefore, a novel in vitro method was used to characterize the scavenging potential of GSH. Accordingly, alkylation of human serum albumin (HSA), which represents an established biomarker for SM intoxication, was used as a measure for remaining SM. Coincubation of GSH and SM in human serum was performed, and time-dependent degradation of SM was monitored in the presence and absence of GSH. Protein-derived and small molecular reaction products between GSH, HSA, and SM were analyzed using microbore liquid chromatography-electrospray ionization high-resolution tandem-mass spectrometry. Although covalent modification of GSH by SM was observed, measurements clearly documented no significant reduction of SM concentration in the presence of GSH. Accordingly, beneficial therapeutic mechanisms of GSH in the case of SM poisoning would appear to be based on physiological effects than on chemical scavenging.


Subject(s)
Antidotes/pharmacology , Chemical Warfare Agents/poisoning , Glutathione/pharmacology , Mustard Gas/poisoning , Alkylation , Biomarkers/analysis , Humans , Hydrolysis , Mass Spectrometry , Serum Albumin, Human/analysis , Serum Albumin, Human/drug effects , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
19.
J Mol Recognit ; 31(4)2018 04.
Article in English | MEDLINE | ID: mdl-29178153

ABSTRACT

In our study, we have characterized the prefibrillar aggregates of human serum albumin (HSA) induced by temsirolimus, anti-renal cancer drug. Molecular docking was retorted to confirm binding of HSA and temsirolimus. Temsirolimus caused the structural transition of native HSA to non-native species after prolonged incubation of 20 days. These non-native species were characterized as prefibrillar aggregates as evident by decreased intrinsic fluorescence and enhanced 8-anilino-1-naphthalene-sulphonic acid (ANS) fluorescence. Further, enhanced thioflavin T fluorescence and shift in congo red (CR) spectra of temsirolimus-incubated HSA as compared to native HSA are suggestive of global transition of HSA in presence of temsirolimus towards prefibrillar aggregates. Circular dichroism spectroscopy revealed α to ß transition upon prolonged incubation with temsirolimus suggesting the formation of prefibrillar aggregates as aggregates are known to possess high ß content. Scanning electron microscopy confirmed these non-native species to be prefibrillar aggregates evident by observed sheath-like structures. Comet assay was retorted to confirm genotoxic nature of these prefibrillar aggregates; DNA damage was observed for temsirolimus-incubated HSA confirming the genotoxic nature of prefibrillar aggregates. These prefibrillar aggregates are observed at heart of many pathological conditions, thus making our study clinically significant.


Subject(s)
Antineoplastic Agents/pharmacology , Serum Albumin, Human/chemistry , Serum Albumin, Human/metabolism , Sirolimus/analogs & derivatives , Antineoplastic Agents/chemistry , Circular Dichroism , Humans , Kidney Neoplasms/drug therapy , Microscopy, Electron, Scanning , Molecular Docking Simulation , Protein Aggregates , Protein Binding , Protein Structure, Secondary , Serum Albumin, Human/drug effects , Sirolimus/chemistry , Sirolimus/pharmacology , Spectrometry, Fluorescence
20.
J Clin Hypertens (Greenwich) ; 19(10): 990-998, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28849629

ABSTRACT

This study investigated the effects and safety of eplerenone or thiazide diuretics in patients with hypertension and albuminuria (pretreatment urinary albumin/creatinine ratio ≥10 mg/gCr) treated with an angiotensin II receptor blocker. The primary end point was the mean percent change in the urinary albumin/creatinine ratio from baseline to 48 weeks. An efficacy analysis was performed in 195 patients (98 in the eplerenone group and 97 in the thiazide group). Systolic and diastolic blood pressures at 48 weeks were similar in the two groups. The mean percent change in the urinary albumin/creatinine ratio from baseline to 48 weeks was similar in the two groups (P=.804). In the safety analysis, the withdrawal rates for adverse events were similar in both groups. The antialbuminuric effects and safety of eplerenone therapy were similar to those of thiazide diuretics when combined with an angiotensin II receptor blocker in patients with hypertension and albuminuria.


Subject(s)
Albuminuria/drug therapy , Hypertension/drug therapy , Serum Albumin, Human/drug effects , Sodium Chloride Symporter Inhibitors/pharmacology , Spironolactone/analogs & derivatives , Aged , Aged, 80 and over , Albuminuria/etiology , Angiotensin Receptor Antagonists/adverse effects , Angiotensin Receptor Antagonists/pharmacology , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Creatinine/blood , Eplerenone , Female , Humans , Male , Middle Aged , Mineralocorticoid Receptor Antagonists/adverse effects , Mineralocorticoid Receptor Antagonists/pharmacology , Prospective Studies , Serum Albumin, Human/urine , Sodium Chloride Symporter Inhibitors/administration & dosage , Sodium Chloride Symporter Inhibitors/adverse effects , Spironolactone/administration & dosage , Spironolactone/adverse effects , Spironolactone/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL